Senolytics decrease senescent cells in humans: Preliminary report from a clinical trial of Dasatinib plus Quercetin in individuals with diabetic kidney disease

LaTonya J Hickson, Larissa G P Langhi Prata, Shane A Bobart, Tamara K Evans, Nino Giorgadze, Shahrukh K Hashmi, Sandra M Herrmann, Michael D Jensen, Qingyi Jia, Kyra L Jordan, Todd A Kellogg, Sundeep Khosla, Daniel M Koerber, Anthony B Lagnado, Donna K Lawson, Nathan K LeBrasseur, Lilach O Lerman, Kathleen M McDonald, Travis J McKenzie, João F Passos, Robert J Pignolo, Tamar Pirtskhalava, Ishran M Saadiq, Kalli K Schaefer, Stephen C Textor, Stella G Victorelli, Tammie L Volkman, Ailing Xue, Mark A Wentworth, Erin O Wissler Gerdes, Yi Zhu, Tamara Tchkonia, James L Kirkland, LaTonya J Hickson, Larissa G P Langhi Prata, Shane A Bobart, Tamara K Evans, Nino Giorgadze, Shahrukh K Hashmi, Sandra M Herrmann, Michael D Jensen, Qingyi Jia, Kyra L Jordan, Todd A Kellogg, Sundeep Khosla, Daniel M Koerber, Anthony B Lagnado, Donna K Lawson, Nathan K LeBrasseur, Lilach O Lerman, Kathleen M McDonald, Travis J McKenzie, João F Passos, Robert J Pignolo, Tamar Pirtskhalava, Ishran M Saadiq, Kalli K Schaefer, Stephen C Textor, Stella G Victorelli, Tammie L Volkman, Ailing Xue, Mark A Wentworth, Erin O Wissler Gerdes, Yi Zhu, Tamara Tchkonia, James L Kirkland

Abstract

Background: Senescent cells, which can release factors that cause inflammation and dysfunction, the senescence-associated secretory phenotype (SASP), accumulate with ageing and at etiological sites in multiple chronic diseases. Senolytics, including the combination of Dasatinib and Quercetin (D + Q), selectively eliminate senescent cells by transiently disabling pro-survival networks that defend them against their own apoptotic environment. In the first clinical trial of senolytics, D + Q improved physical function in patients with idiopathic pulmonary fibrosis (IPF), a fatal senescence-associated disease, but to date, no peer-reviewed study has directly demonstrated that senolytics decrease senescent cells in humans.

Methods: In an open label Phase 1 pilot study, we administered 3 days of oral D 100 mg and Q 1000 mg to subjects with diabetic kidney disease (N = 9; 68·7 ± 3·1 years old; 2 female; BMI:33·9 ± 2·3 kg/m2; eGFR:27·0 ± 2·1 mL/min/1·73m2). Adipose tissue, skin biopsies, and blood were collected before and 11 days after completing senolytic treatment. Senescent cell and macrophage/Langerhans cell markers and circulating SASP factors were assayed.

Findings: D + Q reduced adipose tissue senescent cell burden within 11 days, with decreases in p16INK4A-and p21CIP1-expressing cells, cells with senescence-associated β-galactosidase activity, and adipocyte progenitors with limited replicative potential. Adipose tissue macrophages, which are attracted, anchored, and activated by senescent cells, and crown-like structures were decreased. Skin epidermal p16INK4A+ and p21CIP1+ cells were reduced, as were circulating SASP factors, including IL-1α, IL-6, and MMPs-9 and -12.

Interpretation: "Hit-and-run" treatment with senolytics, which in the case of D + Q have elimination half-lives <11 h, significantly decreases senescent cell burden in humans. FUND: NIH and Foundations. ClinicalTrials.gov Identifier: NCT02848131. Senescence, Frailty, and Mesenchymal Stem Cell Functionality in Chronic Kidney Disease: Effect of Senolytic Agents.

Keywords: Cellular senescence; Dasatinib; Diabetic kidney disease; Quercetin; Senescence-associated secretory phenotype; Senolytics.

Conflict of interest statement

J.L.K., T.T., Y.Z., and N.K.L. have a financial interest related to this research. Patents on senolytic drugs are held by Mayo Clinic. This research has been reviewed by the Mayo Clinic Conflict of Interest Review Board and was conducted in compliance with Mayo Clinic Conflict of Interest policies. No conflicts of interest, financial or otherwise, are declared by the other authors.

Copyright © 2019. Published by Elsevier B.V.

Figures

Fig. 1
Fig. 1
D + Q decreases human adipose tissue senescent cells. (a). D + Q significantly reduced (p = 0·001) abdominal subcutaneous adipose tissue p16INK4A+ cells. Raw values were reduced by 35% in sections of adipose tissue biopsied at Day 14 (11 days after the last dose of a 3-day course of the senolytics) vs. at baseline (Day 0). At Day 0, there were 3·18 ± 0·64 p16INK4A+ cells/100 adipocytes (means ± SEM in 30 fields [400 × 300 μm] at 40× magnification). Means, standard errors, and standard deviations are shown in these “box and whisker” plots. The y axis shows p16INK4A+ cells in the 2 biopsies from each subject at Days 0 and 14 as % of each other (Arbitrary Units). N = 9 subjects; Welch's unpaired 2-tailed t-test for unequal variances. Representative images at Days 0 and 14 are shown. (b). D + Q significantly reduced (p = 0·009) adipose tissue p21CIP1+ cells. Raw values were decreased 17% by 11 days after completing D + Q treatment. At baseline (Day 0), there were 3·82 ± 0·65 p21CIP1+ cells/100 adipocytes (N = 9 subjects; means ± SEM). Representative images are shown. (c). D + Q significantly reduced (p = 0·005) adipose tissue SA-βgal-expressing cells. Raw values were decreased by 62% by 11 days after completing D + Q treatment. At baseline (Day 0), there were 8·76 ± 2·51 SAβgal+ cells/100 nuclei (N = 9 subjects; mean ± SEM). Representative images are shown. Scale bars = 100 μm. Exact p values are indicated. Colours indicate each individual's values on Days 0 and 14.
Fig. 2
Fig. 2
D + Q decreases human adipose tissue macrophages and crown-like structures. (a). D + Q significantly reduced (p = 0·0001) adipose tissue CD68+ macrophages relative to adipocytes. Raw numbers were decreased 28% by 11 days after completing D + Q treatment. At baseline, there were 8·4 ± 0·58 CD68+ macrophages/100 adipocytes (N = 9 subjects; mean ± SEM). Representative images at Days 0 and 14 are shown. (b). D + Q significantly reduced (p = 0·001) adipose tissue crown-like structures. Raw values were decreased by 11 days after D + Q treatment. At baseline (Day 0), there were 0·27 ± 0·05 CD68+ crown-like structures/100 adipocytes (N = 9 subjects; mean ± SEM). Representative images are shown. Scale bars = 100 μm. Exact p values are indicated. Colours indicate each individual's values on Days 0 and 14.
Fig. 3
Fig. 3
Increases in adipocyte progenitor cell density over time are enhanced following administration of D + Q, consistent with removal of cells with limited replicative potential (senescent and pre-senescent cells). Cell density/time was assayed by tetrazolium uptake in adipocyte progenitors isolated from adipose biopsies acquired before (Day 0) and 14 days after the first dose of the 3-day course of D + Q (Day 14) and cultured in parallel for 3 passages. Increases in cell density/time in adipocyte progenitors isolated after senolytic treatment were 8% greater than in adipocyte progenitors isolated before treatment (N = 11 subjects; Table 1). Exact p value is indicated. Colours indicate each individual's values on Days 0 and 14.
Fig. 4
Fig. 4
D + Q decreases human epidermal senescent cells. (a). D + Q significantly reduced (p = 0·026) human epidermal basal layer p16INK4A+ cells. Raw values were decreased 20% by 11 days after completing D + Q treatment. At baseline (Day 0), there were 1·95 ± 0·63 p16INK4A+ cells/mm of epidermis (N = 9 subjects; mean ± SEM). Representative images at Days 0 and 14 are shown. (b). D + Q significantly reduced (p = 0·016) human epidermal basal layer p21CIP1+ cells. Raw values were decreased 31% by 11 days after completing D + Q treatment. At baseline (Day 0), there were 1·71 ± 0·31 p21CIP1+ cells/mm of epidermis (mean ± SEM). Representative images are shown. (c). D + Q did not substantially change (p = .803) antigen-presenting CD1a+ epidermal Langerhans immune cells. At baseline (Day 0), there were 14·55 ± 2·16 CD1a+ cells/mm of epidermis (mean ± SEM; N = 9). Scale bars = 100 μm. Exact p values are indicated. Colours indicate each individual's values on Days 0 and 14.
Fig. 5
Fig. 5
D + Q decreases plasma SASP factors. Plasma SASP factors were assayed at baseline (Day 0) and after treatment (Day 14). Key SASP factors that were significantly decreased (p < 0·05) after treatment are shown. Colours indicate each individual's values on Days 0 and 14.

References

    1. Hayflick L., Moorehead P. The serial cultivation of human diploid strains. Exp Cell Res. 1961;25:585–621.
    1. Kirkland J.L., Tchkonia T. Cellular senescence: a translational perspective. EBioMedicine. 2017;21:21–28.
    1. Lewis-McDougall F.C., Ruchaya P.J., Domenjo-Vila E., Shin Teoh T., Prata L., Cottle B.J. Aged-senescent cells contribute to impaired heart regeneration. Aging Cell. 2019;18
    1. Kirkland J.L., Hollenberg C.H., Gillon W.S. Age, anatomic site, and the replication and differentiation of adipocyte precursors. Am J Physiol. 1990;258 [C206-C10]
    1. Farr J.N., Xu M., Weivoda M.M., Monroe D.G., Fraser D.G., Onken J.L. Targeting cellular senescence prevents age-related bone loss in mice. Nat Med. 2017:231072–231079.
    1. Justice J.N., Gregory H., Tchkonia T., LeBrasseur N.K., Kirkland J.L., Kritchevsky S.B. Cellular senescence biomarker p16INK4a+ cell burden in thigh adipose is associated with poor physical function in older women. T J Gerontol A Biol Sci Med Sci. 2018;73:939–945.
    1. Ogrodnik M., Miwa S., Tchkonia T., Tiniakos D., Wilson C.L., Lahat A. Cellular senescence drives age-dependent hepatic steatosis. Nat Commun. 2017 Jun 13;8:15691.
    1. Farr J.N., Fraser D.G., Wang H., Jaehn K., Ogrodnik M.B., Weivoda M.M. Identification of senescent cells in the bone microenvironment. J Bone Miner Res. 2016;31:1920–1929.
    1. Roos C.M., Zhang B., Palmer A.K., Ogrodnik M.B., Pirtskhalava T., Thalji N.M. Chronic senolytic treatment alleviates established vasomotor dysfunction in aged or atherosclerotic mice. Aging Cell. 2016;15:973–977.
    1. Xu M., Palmer A.K., Ding H., Weivoda M.M., Pirtskhalava T., White T.A. Targeting senescent cells enhances adipogenesis and metabolic function in old age. eLife. 2015
    1. Xu M., Tchkonia T., Ding H., Ogrodnik M., Lubbers E.R., Pirtskhalava T. JAK inhibition alleviates the cellular senescence-associated secretory phenotype and frailty in old age. P Proc Natl Acad Sci (USA) 2015;112 [E6301-E10]
    1. Zhu Y., Armstrong J.L., Tchkonia T., Kirkland J.L. Cellular senescence and the senescent secretory phenotype in age-related chronic diseases. Curr Opin Clin Nutr Metab Care. 2014;17:324–328.
    1. Tchkonia T., Morbeck D.E., von Zglinicki T., van Deursen J., Lustgarten J., Scrable H. Fat tissue, aging, and cellular senescence. Aging Cell. 2010;9:667–684.
    1. Kirkland J.L., Dobson D.E. Preadipocyte function and aging: links between age-related changes in cell dynamics and altered fat cell function. J Amer Geriatr Soc. 1997;45:959–967.
    1. Tchkonia T., Giorgadze N., Pirtskhalava T., Thomou T., Villaret A., Bouloumie A. Cellular senescence and inflammation in obesity. Obesity. 2009;17(Suppl. 2):S57.
    1. Villaret A., Galitzky J., Decaunes P., Esteve D., Marques M.A., Sengenes C. Adipose tissue endothelial cells from obese human subjects: differences among depots in angiogenic, metabolic, and inflammatory gene expression and cellular senescence. Diabetes. 2010;59:2755–2763.
    1. Minamino T., Orimo M., Shimizu I., Kunieda T., Yokoyama M., Ito T. A crucial role for adipose tissue p53 in the regulation of insulin resistance. N Nat Med. 2009;15:1082–1087.
    1. Docherty M.H., O'Sullivan E.D., Bonventre J.V., Ferenbach D.A. Cellular senescence in the kidney. J Am Soc Nephrol. 2019;30:726–736.
    1. Dai L., Qureshi A.R., Witasp A., Lindholm B., Stenvinkel P. Early vascular ageing and cellular senescence in chronic kidney disease. Comput Struct Biotechnol J. 2019;17:721–729.
    1. Kim S.R., Jiang K., Ogrodnik M., Chen X., Zhu X.Y., Lohmeier H. Increased renal cellular senescence in murine high-fat diet: effect of the senolytic drug quercetin. Transl Res. 2019
    1. Kirkland J.L., Tchkonia T., Zhu Y., Niedernhofer L.J., Robbins P.D. The clinical potential of senolytic drugs. J Amer Geriatr Soc. 2017 Oct;65(10):2297–2301.
    1. Tchkonia T., Kirkland J.L. Aging, cell senescence, and chronic disease: emerging therapeutic strategies. JAMA. 2018;320 [1319–200]
    1. Palmer A.K., Xu M., Zhu Y., Pirtskhalava T., Weivoda M.M., Hachfeld C.M. Targeting senescent cells alleviates obesity-induced metabolic dysfunction. Aging Cell. 2019;18
    1. Ogrodnik M., Zhu Y., Langhi L.G.P., Tchkonia T., Kruger P., Fielder E. Obesity-induced cellular senescence drives anxiety and impairs neurogenesis. Cell Metab. 2019;29:1061–1077.
    1. Musi N., Valentine J.M., Sickora K.R., Baeuerle E., Thompson C.S., Shen Q. Tau protein aggregation is associated with cellular senescence in the brain. Aging Cell. 2018;17
    1. Coppé J.P., Patil C., Rodier F., Sun Y., Muñoz D.P., Goldstein J. Senescence-associated secretory phenotypes reveal cell-nonautonomous functions of oncogenic RAS and the p53 tumor suppressor. PLoS Biol. 2008;6:2853–2868.
    1. Xu M., Bradley E.W., Weivoda M.M., Hwang S.M., Pirtskhalava T., Decklever T. Transplanted senescent cells induce an osteoarthritis-like condition in mice. J Gerontol A Biol Sci Med Sci. 2017;72:780–785.
    1. Xu M., Pirtskhalava T., Farr J.N., Weigand B.M., Palmer A.K., Weivoda M.M. Senolytics improve physical function and increase lifespan in old age. Nat Med. 2018;24:1246–1256.
    1. Wang E. Senescent human fibroblasts resist programmed cell death, and failure to suppress BCL2 is involved. Cancer Res. 1995;55:2284–2292.
    1. Kirkland J.L., Tchkonia T. Clinical strategies and animal models for developing senolytic agents. Exp Gerontol. 2015;68:19–25.
    1. Krishnamurthy J., Torrice C., Ramsey M.R., Kovalev G.I., Al-Regaiey K., Su L. Ink4a/Arf expression is a biomarker of aging. J Clin Invest. 2004;114:1299–1307.
    1. Zhu Y., Tchkonia T., Pirtskhalava T., Gower A., Ding H., Giorgadze N. The Achilles' heel of senescent cells: from transcriptome to senolytic drugs. Aging Cell. 2015;14:644–658.
    1. Schafer M.J., White T.A., Iijima K., Haak A.J., Ligresti G., Atkinson E.J. Cellular senescence mediates fibrotic pulmonary disease. Nat Commun. 2017;8:14532.
    1. Parikh P., Britt R.D., Jr., Manlove L.J., Wicher S.A., Roesler A., Ravix J. Hyperoxia-induced cellular senescence in fetal airway smooth muscle cells. Am J Respir Cell Mol Biol. 2019;61:51–60.
    1. Nath K.A., O'Brien D.R., Croatt A.J., Grande J.P., Ackerman A.W., Nath M.C. The murine dialysis fistula model exhibits a senescence phenotype: pathobiologic mechanisms and therapeutic potential. Am J Physiol Renal Physiol. 2018;315:F1493–F1499.
    1. Zhang P., Kishimoto Y., Grammatikakis I., Gottimukkala K., Cutler R.G., Zhang S. Senolytic therapy alleviates Abeta-associated oligodendrocyte progenitor cell senescence and cognitive deficits in an Alzheimer's disease model. Nat Neurosci. 2019;22:719–728.
    1. Fuhrmann-Stroissnigg H., Ling Y.Y., Zhao J., McGowan S.J., Stripay J.L., Gregg S. Identification of HSP90 inhibitors as senolytics for extending healthspan. Nat Commun. 2017;8:422.
    1. Christopher L.J., Cui D., Wu C., Luo R., Manning J.A., Bonacorsi S.J. Metabolism and disposition of dasatinib after oral administration to humans. Drug Metab Dispos. 2008;36:1357–1364.
    1. Graefe E.U., Wittig J., Mueller S., Riethling A.K., Uehleke B., Drewelow B. Pharmacokinetics and bioavailability of quercetin glycosides in humans. J Clin Pharmacol. 2001;41:492–499.
    1. Justice J.N., Nambiar A.M., Tchkonia T., LeBrasseur N.K., Pascual R., Hashmi S.K. Senolytics in idiopathic pulmonary fibrosis: results from a first-in-human, open-label, pilot study. EBioMedicine. 2019;40:554–563.
    1. Martyanov V., Whitfield M.L., Varga J. Senescence signature in skin biopsies from systemic sclerosis patients treated with senolytic therapy: potential predictor of clinical response? Arthritis Rheumatol. 2019
    1. Verzola D., Gandolfo M.T., Gaetani G., Ferraris A., Mangerini R., Ferrario F. Accelerated senescence in the kidneys of patients with type 2 diabetic nephropathy. Am J Physiol Renal Physiol. 2008;295(5):F1563–F1573.
    1. Liu J., Yang J.R., Chen X.M., Cai G.Y., Lin L.R., He Y.N. Impact of ER stress-regulated ATF4/p16 signaling on the premature senescence of renal tubular epithelial cells in diabetic nephropathy. Am J Physiol Cell Physiol. 2015;308:C621–C630.
    1. Satriano J., Mansoury H., Deng A., Sharma K., Vallon V., Blantz R.C. Transition of kidney tubule cells to a senescent phenotype in early experimental diabetes. Am J Physiol Cell Physiol. 2010;299:C374–C380.
    1. Schneider C.A., Rasband W.S., Eliceiri K.W. NIH image to ImageJ: 25 years of image analysis. Nat Methods. 2012;9:671–675.
    1. Schindelin J., Arganda-Carreras I., Frise E., Kaynig V., Longair M., Pietzsch T. Fiji: an open-source platform for biological-image analysis. Nat Methods. 2012;9:676–682.
    1. Morgan-Bathke M., Harteneck D., Jaeger P., Sondergaard E., Karwoski R., Espinosa De Ycaza A. Comparison of methods for analyzing human adipose tissue macrophage content. Obesity. 2017;25:2100–2107.
    1. Eirin A., Zhu X.Y., Krier J.D., Tang H., Jordan K.L., Grande J.P. Adipose tissue-derived mesenchymal stem cells improve revascularization outcomes to restore renal function in swine atherosclerotic renal artery stenosis. Stem Cells. 2012;30:1030–1041.
    1. Saad A., Dietz A.B., Herrmann S.M.S., Hickson L.J., Glockner J.F., McKusick M.A. Autologous mesenchymal stem cells increase cortical perfusion in renovascular disease. J Am Soc Nephrol. 2017;28:2777–2785.
    1. Wang A.S., Dreesen O. Biomarkers of cellular senescence and skin aging. Front Genet. 2018;9:247.
    1. Bernardes de Jesus B., Blasco M.A. Assessing cell and organ senescence biomarkers. Circ Res. 2012;111:97–109.
    1. Murano I., Barbatelli G., Parisani V., Latini C., Muzzonigro G., Castellucci M. Dead adipocytes, detected as crown-like structures, are prevalent in visceral fat depots of genetically obese mice. J Lipid Res. 2008;49:1562–1568.
    1. Mitsui Y., Schneider E.L. Relationship between cell replication and volume in senescent human diploid fibroblasts. Mech Ageing Dev. 1976;5:45–56.
    1. Schneider E.L. Cell replication and aging: in vitro and in vivo studies. Fed Proc. 1979;38:1857–1861.
    1. Zhu Y., Tchkonia T., Stout M.B., Giorgadze N., Wang L., Li P.W. Inflammation and the depot-specific secretome of human preadipocytes. Obesity. 2015;23:989–999.
    1. Tchkonia T., Cartwright M., Wise B., Pirtskhalava T., Karagiannides I., Shpilman A. Increased TNFa and CCAAT/enhancer binding protein homologous protein (CHOP) with aging predispose preadipocytes to resist adipogenesis. Am J Physiol. 2007;293 [E1810-E9]
    1. Tchkonia T., Lenburg M., Thomou T., Giorgadze N., Frampton G., Pirtskhalava T. Identification of depot-specific human fat cell progenitors through distinct expression profiles and developmental gene patterns. Am J Physiol. 2007;292:E298–E307.
    1. Doebel T., Voisin B., Nagao K. Langerhans cells - the macrophage in dendritic cell clothing. Trends Immunol. 2017;38:817–828.
    1. Hall B.M., Balan V., Gleiberman A.S., Strom E., Krasnov P., Virtuoso L.P. p16(Ink4a) and senescence-associated beta-galactosidase can be induced in macrophages as part of a reversible response to physiological stimuli. Aging (Albany NY) 2017;9:1867–1884.
    1. Waaijer M.E., Parish W.E., Strongitharm B.H., van Heemst D., Slagboom P.E., de Craen A.J. The number of p16INK4a positive cells in human skin reflects biological age. Aging Cell. 2012;11:722–725.
    1. Bhalla S., Tremblay D., Mascarenhas J. Discontinuing tyrosine kinase inhibitor therapy in chronic myelogenous leukemia: current understanding and future directions. Clin Lymphoma Myeloma Leuk. 2016;16:488–494.
    1. Fox L.C., Cummins K.D., Costello B., Yeung D., Cleary R., Forsyth C. The incidence and natural history of dasatinib complications in the treatment of chronic myeloid leukemia. Blood Adv. 2017;1:802–811.
    1. La Rosee P., Martiat P., Leitner A., Klag T., Muller M.C., Erben P. Improved tolerability by a modified intermittent treatment schedule of dasatinib for patients with chronic myeloid leukemia resistant or intolerant to imatinib. Ann Hematol. 2013;92:1345–1350.
    1. Zhu Y., Doornebal E.J., Pirtskhalava T., Giorgadze N., Wentworth M., Fuhrmann-Stroissnigg H. New agents that target senescent cells: the flavone, fisetin, and the BCL-XL inhibitors, A1331852 and A1155463. Aging (Albany NY) 2017;9:955–963.
    1. Yousefzadeh M.J., Zhu Y., McGowan S.J., Angelini L., Fuhrmann-Stroissnigg H., Xu M. Fisetin is a senotherapeutic that extends health and lifespan. EBioMedicine. 2018 Oct;36:18–28.
    1. Wang Y., Chang J., Liu X., Zhang X., Zhang S., Zhou D. Discovery of piperlongumine as a potential novel lead for the development of senolytic agents. Aging (Milano) 2016;8:2915–2926.
    1. Ozsvari B., Nuttall J.R., Sotgia F., Lisanti M.P. Azithromycin and Roxithromycin define a new family of "senolytic" drugs that target senescent human fibroblasts. Aging (Milano) 2018;10:3294–3307.
    1. Cherif H., Bisson D.G., Jarzem P., Weber M., Ouellet J.A., Haglund L. Curcumin and o-vanillin exhibit evidence of senolytic activity in human IVD cells in vitro. J Clin Med. 2019;8(4) pii: E433.
    1. Li W., He Y., Zhang R., Zheng G., Zhou D. The curcumin analog EF24 is a novel senolytic agent. Aging (Milano) 2019;11:771–782.
    1. Samaraweera L., Adomako A., Rodriguez-Gabin A., McDaid H.M. A novel indication for panobinostat as a senolytic drug in NSCLC and HNSCC. Sci Rep. 2017;7:1900.
    1. Zhu Y., Tchkonia T., Fuhrmann-Stroissnigg H., Dai H.M., Ling Y.Y., Stout M.B. Identification of a novel senolytic agent, navitoclax, targeting the Bcl-2 family of anti-apoptotic factors. Aging Cell. 2015;14:644–658.
    1. Kaefer A., Yang J., Noertersheuser P., Mensing S., Humerickhouse R., Awni W. Mechanism-based pharmacokinetic/pharmacodynamic meta-analysis of navitoclax (ABT-263) induced thrombocytopenia. Cancer Chemother Pharmacol. 2014;74:593–602.
    1. Mahfoudhi E., Lordier L., Marty C., Pan J., Roy A., Roy L. P53 activation inhibits all types of hematopoietic progenitors and all stages of megakaryopoiesis. Oncotarget. 2016;7:31980–31992.
    1. Justice J., Miller J.D., Newman J.C., Hashmi S.K., Halter J., Austad S.N. Frameworks for proof-of-concept clinical trials of interventions that target fundamental aging processes. J Gerontol A Biol Sci Med Sci. 2016;71:1415–1423.
    1. Kennedy B.K., Berger S.L., Brunet A., Campisi J., Cuervo A.M., Epel E.S. Geroscience: linking aging to chronic disease. Cell. 2014;159:709–713.
Web Reference 1
    1. ,

Source: PubMed

3
Tilaa